1
|
Tang P, Wei F, Qiao W, Chen X, Ji C, Yang W, Zhang X, Chen S, Wu Y, Jiang M, Ma C, Shen W, Dong Q, Cao H, Xie M, Cai Z, Xu L, Shi J, Dong N, Chen J, Wang N. Engineering aortic valves via transdifferentiating fibroblasts into valvular endothelial cells without using viruses or iPS cells. Bioact Mater 2025; 45:181-200. [PMID: 39651397 PMCID: PMC11625219 DOI: 10.1016/j.bioactmat.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The technology of induced pluripotent stem cells (iPSCs) has enabled the conversion of somatic cells into primitive undifferentiated cells via reprogramming. This approach provides possibilities for cell replacement therapies and drug screening, but the potential risk of tumorigenesis hampers its further development and in vivo application. How to generate differentiated cells such as valvular endothelial cells (VECs) has remained a major challenge. Utilizing a combinatorial strategy of selective soluble chemicals, cytokines and substrate stiffness modulation, mouse embryonic fibroblasts are directly and efficiently transdifferentiated into induced aortic endothelial cell-like cells (iAECs), or human primary adult fibroblasts are transdifferentiated into induced valvular endothelial cell-like cells (hiVECs), without expressing pluripotency stem cell markers. These iAECs and hiVECs express VEC-associated genes and proteins and VEC-specific marker NFATC1 and are functional in culture and on decellularized porcine aortic valves, like mouse aortic endothelial cells or human primary aortic valvular endothelial cells. The iAECs and hiVECs seeded on decellularized porcine aortic valves stay intact and express VEC-associated proteins for 60 days after grafting into abdominal aorta of immune-compromised rats. In contrast, induced pluripotent stem cells (iPSCs) are less efficient in differentiating into VEC-like cells and pluripotency marker Nanog is expressed in a small subpopulation of iPSC-derived VEC-like cells that generate teratomas in SCID mice whereas hiVECs derived from transdifferentiation do not generate teratomas in vivo. Our findings highlight an approach to efficiently convert fibroblasts into iAECs and hiVECs and seed them onto decellularized aortic valves for safely generating autologous tissue-engineered aortic valves without using viruses or first reprogramming the cells into pluripotent stem cells.
Collapse
Affiliation(s)
- Peng Tang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Fuxiang Wei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chenyang Ji
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Wanzhi Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Xinyu Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Sihan Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yanyan Wu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Mingxing Jiang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Chenyu Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Weiqiang Shen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Qi Dong
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Minghui Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ziwen Cai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junwei Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Ning Wang
- Institute for Mechanobiology, Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
2
|
Ba T, Miao H, Zhang L, Gao C, Wang Y. ClusterMatch aligns single-cell RNA-sequencing data at the multi-scale cluster level via stable matching. Bioinformatics 2024; 40:btae480. [PMID: 39073888 PMCID: PMC11520419 DOI: 10.1093/bioinformatics/btae480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/04/2024] [Accepted: 07/28/2024] [Indexed: 07/31/2024] Open
Abstract
MOTIVATION Unsupervised clustering of single-cell RNA sequencing (scRNA-seq) data holds the promise of characterizing known and novel cell type in various biological and clinical contexts. However, intrinsic multi-scale clustering resolutions poses challenges to deal with multiple sources of variability in the high-dimensional and noisy data. RESULTS We present ClusterMatch, a stable match optimization model to align scRNA-seq data at the cluster level. In one hand, ClusterMatch leverages the mutual correspondence by canonical correlation analysis and multi-scale Louvain clustering algorithms to identify cluster with optimized resolutions. In the other hand, it utilizes stable matching framework to align scRNA-seq data in the latent space while maintaining interpretability with overlapped marker gene set. Through extensive experiments, we demonstrate the efficacy of ClusterMatch in data integration, cell type annotation, and cross-species/timepoint alignment scenarios. Our results show ClusterMatch's ability to utilize both global and local information of scRNA-seq data, sets the appropriate resolution of multi-scale clustering, and offers interpretability by utilizing marker genes. AVAILABILITY AND IMPLEMENTATION The code of ClusterMatch software is freely available at https://github.com/AMSSwanglab/ClusterMatch.
Collapse
Affiliation(s)
- Teer Ba
- School of Physical Science and Technology, Inner Mongolia University, Hohhot 010021, China
- School of Mathematical Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Hao Miao
- CEMS, NCMIS, HCMS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Lirong Zhang
- School of Physical Science and Technology, Inner Mongolia University, Hohhot 010021, China
| | - Caixia Gao
- School of Mathematical Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Yong Wang
- CEMS, NCMIS, HCMS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 330106, China
| |
Collapse
|
3
|
Deng D, Zhang Y, Tang B, Zhang Z. Sources and applications of endothelial seed cells: a review. Stem Cell Res Ther 2024; 15:175. [PMID: 38886767 PMCID: PMC11184868 DOI: 10.1186/s13287-024-03773-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
Endothelial cells (ECs) are widely used as donor cells in tissue engineering, organoid vascularization, and in vitro microvascular model development. ECs are invaluable tools for disease modeling and drug screening in fundamental research. When treating ischemic diseases, EC engraftment facilitates the restoration of damaged blood vessels, enhancing therapeutic outcomes. This article presents a comprehensive overview of the current sources of ECs, which encompass stem/progenitor cells, primary ECs, cell lineage conversion, and ECs derived from other cellular sources, provides insights into their characteristics, potential applications, discusses challenges, and explores strategies to mitigate these issues. The primary aim is to serve as a reference for selecting suitable EC sources for preclinical research and promote the translation of basic research into clinical applications.
Collapse
Affiliation(s)
- Dan Deng
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yu Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Bo Tang
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
4
|
Zhang L, Wu X, Hong L. Endothelial Reprogramming in Atherosclerosis. Bioengineering (Basel) 2024; 11:325. [PMID: 38671747 PMCID: PMC11048243 DOI: 10.3390/bioengineering11040325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Atherosclerosis (AS) is a severe vascular disease that results in millions of cases of mortality each year. The development of atherosclerosis is associated with vascular structural lesions, characterized by the accumulation of immune cells, mesenchymal cells, lipids, and an extracellular matrix at the intimal resulting in the formation of an atheromatous plaque. AS involves complex interactions among various cell types, including macrophages, endothelial cells (ECs), and smooth muscle cells (SMCs). Endothelial dysfunction plays an essential role in the initiation and progression of AS. Endothelial dysfunction can encompass a constellation of various non-adaptive dynamic alterations of biology and function, termed "endothelial reprogramming". This phenomenon involves transitioning from a quiescent, anti-inflammatory state to a pro-inflammatory and proatherogenic state and alterations in endothelial cell identity, such as endothelial to mesenchymal transition (EndMT) and endothelial-to-immune cell-like transition (EndIT). Targeting these processes to restore endothelial balance and prevent cell identity shifts, alongside modulating epigenetic factors, can attenuate atherosclerosis progression. In the present review, we discuss the role of endothelial cells in AS and summarize studies in endothelial reprogramming associated with the pathogenesis of AS.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Xin Wu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Liang Hong
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Huang NF, Stern B, Oropeza BP, Zaitseva TS, Paukshto MV, Zoldan J. Bioengineering Cell Therapy for Treatment of Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:e66-e81. [PMID: 38174560 PMCID: PMC10923024 DOI: 10.1161/atvbaha.123.318126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Peripheral artery disease is an atherosclerotic disease associated with limb ischemia that necessitates limb amputation in severe cases. Cell therapies comprised of adult mononuclear or stromal cells have been clinically tested and show moderate benefits. Bioengineering strategies can be applied to modify cell behavior and function in a controllable fashion. Using mechanically tunable or spatially controllable biomaterials, we highlight examples in which biomaterials can increase the survival and function of the transplanted cells to improve their revascularization efficacy in preclinical models. Biomaterials can be used in conjunction with soluble factors or genetic approaches to further modulate the behavior of transplanted cells and the locally implanted tissue environment in vivo. We critically assess the advances in bioengineering strategies such as 3-dimensional bioprinting and immunomodulatory biomaterials that can be applied to the treatment of peripheral artery disease and then discuss the current challenges and future directions in the implementation of bioengineering strategies.
Collapse
Affiliation(s)
- Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Brett Stern
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | | | | | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| |
Collapse
|
6
|
Kizub IV. Induced pluripotent stem cells for cardiovascular therapeutics: Progress and perspectives. REGULATORY MECHANISMS IN BIOSYSTEMS 2023; 14:451-468. [DOI: 10.15421/10.15421/022366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The discovery of methods for reprogramming adult somatic cells into induced pluripotent stem cells (iPSCs) opens up prospects of developing personalized cell-based therapy options for a variety of human diseases as well as disease modeling and new drug discovery. Like embryonic stem cells, iPSCs can give rise to various cell types of the human body and are amenable to genetic correction. This allows usage of iPSCs in the development of modern therapies for many virtually incurable human diseases. The review summarizes progress in iPSC research in the context of application in the cardiovascular field including modeling cardiovascular disease, drug study, tissue engineering, and perspectives for personalized cardiovascular medicine.
Collapse
|
7
|
Cho S, Aakash P, Lee S, Yoon YS. Endothelial cell direct reprogramming: Past, present, and future. J Mol Cell Cardiol 2023; 180:22-32. [PMID: 37080451 PMCID: PMC10330356 DOI: 10.1016/j.yjmcc.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Ischemic cardiovascular disease still remains as a leading cause of morbidity and mortality despite various medical, surgical, and interventional therapy. As such, cell therapy has emerged as an attractive option because it tackles underlying problem of the diseases by inducing neovascularization in ischemic tissue. After overall failure of adult stem or progenitor cells, studies attempted to generate endothelial cells (ECs) from pluripotent stem cells (PSCs). While endothelial cells (ECs) differentiated from PSCs successfully induced vascular regeneration, differentiating volatility and tumorigenic potential is a concern for their clinical applications. Alternatively, direct reprogramming strategies employ lineage-specific factors to change cell fate without achieving pluripotency. ECs have been successfully reprogrammed via ectopic expression of transcription factors (TFs) from endothelial lineage. The reprogrammed ECs induced neovascularization in vitro and in vivo and thus demonstrated their therapeutic value in animal models of vascular insufficiency. Methods of delivering reprogramming factors include lentiviral or retroviral vectors and more clinically relevant, non-integrative adenoviral and episomal vectors. Most studies made use of fibroblast as a source cell for reprogramming, but reprogrammability of other clinically relevant source cell types has to be evaluated. Specific mechanisms and small molecules that are involved in the aforementioned processes tackles challenges associated with direct reprogramming efficiency and maintenance of reprogrammed EC characteristics. After all, this review provides summary of past and contemporary methods of direct endothelial reprogramming and discusses the future direction to overcome these challenges to acquire clinically applicable reprogrammed ECs.
Collapse
Affiliation(s)
- Seonggeon Cho
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Parthasarathy Aakash
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Sangho Lee
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| | - Young-Sup Yoon
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Turyova E, Mikolajcik P, Grendar M, Kudelova E, Holubekova V, Kalman M, Marcinek J, Hrnciar M, Kovac M, Miklusica J, Laca L, Lasabova Z. Expression of OCT4 isoforms is reduced in primary colorectal cancer. Front Oncol 2023; 13:1166835. [PMID: 37409260 PMCID: PMC10319064 DOI: 10.3389/fonc.2023.1166835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/02/2023] [Indexed: 07/07/2023] Open
Abstract
Introduction Colorectal cancer (CRC) is one of the most common types of cancer worldwide. The carcinogenesis of CRC is indeed complex, and there are many different mechanisms and pathways that contribute to the development of malignancy and the progression from primary to metastatic tumors. The OCT4A, encoded by the POU5F1 gene, is a transcription factor responsible for the phenotype of stem cells, maintaining pluripotency and regulation of differentiation. The POU5F1 gene is made up of five exons that can create numerous isoforms through alternative promoter or alternative splicing. In addition to OCT4A, other isoforms called OCT4B are also translated into protein; however, their role in cells has been unclear. The aim of our work was to investigate the expression patterns of OCT4 isoforms in primary and metastatic CRC, providing us with useful information about their role in the development and progression of CRC. Methods Surgical specimens from a total of 78 patients were collected and isolated from primary tumors (n = 47) and metastases (n = 31). The relative gene expression of OCT4 isoforms was investigated using the RT-qPCR method together with the TaqMan probes for particular OCT4 isoforms. Results Our results suggest significantly downregulated expression of the OCT4A and OCT4Bs isoforms in both primary (p = 0.0002 and p < 0.0001, respectively) and metastatic tumors (p = 0.0006 and p = 0.00051, respectively) when compared with the control samples. We also observed a correlation between reduced expression of all OCT4 isoforms and both primary and left-sided tumors (p = 0.001 and p = 0.030, respectively). On the other hand, the expression of all OCT4 isoforms was significantly upregulated in metastases compared with primary tumors (p < 0.0001). Discussion Unlike previous reports, we found out that the expression of OCT4A, OCT4Bs, and all OCT4 isoforms was significantly reduced in primary tumors and metastases compared with control samples. On the other hand, we supposed that the expression rate of all OCT4 isoforms may be related to the cancer type and side, as well as to liver metastases. However, further studies are required to investigate the detailed expression patterns and significance of individual OCT4 isoforms in carcinogenesis.
Collapse
Affiliation(s)
- Eva Turyova
- Department of Molecular Biology and Genomics, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Martin, Slovakia
| | - Peter Mikolajcik
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin and University Hospital Martin, Comenius University Bratislava, Martin, Slovakia
| | - Marian Grendar
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Martin, Slovakia
| | - Eva Kudelova
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin and University Hospital Martin, Comenius University Bratislava, Martin, Slovakia
| | - Veronika Holubekova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Martin, Slovakia
| | - Michal Kalman
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin and University Hospital Martin, Comenius University Bratislava, Martin, Slovakia
| | - Juraj Marcinek
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin and University Hospital Martin, Comenius University Bratislava, Martin, Slovakia
| | - Matej Hrnciar
- Department of Informatics, Information Systems and Software Engineering, Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Michal Kovac
- Department of Informatics, Information Systems and Software Engineering, Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Juraj Miklusica
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin and University Hospital Martin, Comenius University Bratislava, Martin, Slovakia
| | - Ludovit Laca
- Clinic of Surgery and Transplant Center, Jessenius Faculty of Medicine in Martin and University Hospital Martin, Comenius University Bratislava, Martin, Slovakia
| | - Zora Lasabova
- Department of Molecular Biology and Genomics, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Martin, Slovakia
| |
Collapse
|
9
|
Chan AH, Hu C, Chiang GC, Ekweume C, Huang NF. Chronic nicotine impairs the angiogenic capacity of human induced pluripotent stem cell-derived endothelial cells in a murine model of peripheral arterial disease. JVS Vasc Sci 2023; 4:100115. [PMID: 37519333 PMCID: PMC10372313 DOI: 10.1016/j.jvssci.2023.100115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/05/2023] [Indexed: 08/01/2023] Open
Abstract
Objective Lifestyle choices such as tobacco and e-cigarette use are a risk factor for peripheral arterial disease (PAD) and may influence therapeutic outcomes. The effect of chronic nicotine exposure on the angiogenic capacity of human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) was assessed in a murine model of PAD. Methods Mice were exposed to nicotine or phosphate-buffered saline (PBS) for 28 days, followed by induction of limb ischemia and iPSC-EC transplantation. Cells were injected into the ischemic limb immediately after induction of hindlimb ischemia and again 7 days later. Limb perfusion was assessed by laser Doppler spectroscopy, and transplant cell survival was monitored for 14 days afterward using bioluminescence imaging, followed by histological analysis of angiogenesis. Results Transplant cell retention progressively decreased over time after implantation based on bioluminescence imaging, and there were no significant differences in cell survival between mice with chronic exposure to nicotine or PBS. However, compared with mice without nicotine exposure, mice with prior nicotine exposure had had an impaired therapeutic response to iPSC-EC therapy based on decreased vascular perfusion recovery. Mice with nicotine exposure, followed by cell transplantation, had significantly lower mean perfusion ratio after 14 days (0.47 ± 0.07) compared with mice undergoing cell transplantation without prior nicotine exposure (0.79 ± 0.11). This finding was further supported by histological analysis of capillary density, in which animals with prior nicotine exposure had a lower capillary density (45.9 ± 4.7 per mm2) compared with mice without nicotine exposure (66.5 ± 8.1 per mm2). Importantly, the ischemic limbs mice exposed to nicotine without cell therapy also showed significant impairment in perfusion recovery after 14 days, compared with mice that received PBS + iPSC-EC treatment. This result suggested that mice without chronic nicotine exposure could respond to iPSC-EC implantation into the ischemic limb by inducing perfusion recovery, whereas mice with chronic nicotine exposure did not respond to iPSC-EC therapy. Conclusions Together, these findings show that chronic nicotine exposure adversely affects the ability of iPSC-EC therapy to promote vascular perfusion recovery and angiogenesis in a murine PAD model.
Collapse
Affiliation(s)
- Alex H.P. Chan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
| | - Caroline Hu
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
| | - Gladys C.F. Chiang
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
| | - Chisomaga Ekweume
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
- College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA
- Department of Chemical Engineering, Stanford University, Stanford, CA
| |
Collapse
|
10
|
Lv J, Meng S, Gu Q, Zheng R, Gao X, Kim JD, Chen M, Xia B, Zuo Y, Zhu S, Zhao D, Li Y, Wang G, Wang X, Meng Q, Cao Q, Cooke JP, Fang L, Chen K, Zhang L. Epigenetic landscape reveals MECOM as an endothelial lineage regulator. Nat Commun 2023; 14:2390. [PMID: 37185814 PMCID: PMC10130150 DOI: 10.1038/s41467-023-38002-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
A comprehensive understanding of endothelial cell lineage specification will advance cardiovascular regenerative medicine. Recent studies found that unique epigenetic signatures preferentially regulate cell identity genes. We thus systematically investigate the epigenetic landscape of endothelial cell lineage and identify MECOM to be the leading candidate as an endothelial cell lineage regulator. Single-cell RNA-Seq analysis verifies that MECOM-positive cells are exclusively enriched in the cell cluster of bona fide endothelial cells derived from induced pluripotent stem cells. Our experiments demonstrate that MECOM depletion impairs human endothelial cell differentiation, functions, and Zebrafish angiogenesis. Through integrative analysis of Hi-C, DNase-Seq, ChIP-Seq, and RNA-Seq data, we find MECOM binds enhancers that form chromatin loops to regulate endothelial cell identity genes. Further, we identify and verify the VEGF signaling pathway to be a key target of MECOM. Our work provides important insights into epigenetic regulation of cell identity and uncovered MECOM as an endothelial cell lineage regulator.
Collapse
Affiliation(s)
- Jie Lv
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Shu Meng
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Qilin Gu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Rongbin Zheng
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Xinlei Gao
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Jun-Dae Kim
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Min Chen
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Bo Xia
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Yihan Zuo
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Sen Zhu
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Dongyu Zhao
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Yanqiang Li
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Xin Wang
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Qingshu Meng
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qi Cao
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
| | - Kaifu Chen
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| | - Lili Zhang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Marzoog BA. Endothelial cell autophagy in the context of disease development. Anat Cell Biol 2023; 56:16-24. [PMID: 36267005 PMCID: PMC9989784 DOI: 10.5115/acb.22.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/27/2022] Open
Abstract
Endothelial cells (EC) are the anatomical boundaries between the intravascular and extravascular space. Damage to ECs is catastrophic and induces endothelial cell dysfunction. The pathogenesis is multifactorial and involves dysregulation in the signaling pathways, membrane lipids ratio disturbance, cell-cell adhesion disturbance, unfolded protein response, lysosomal and mitochondrial stress, autophagy dysregulation, and oxidative stress. Autophagy is a lysosomal-dependent turnover of intracellular components. Autophagy was recognized early in the pathogenesis of endothelial dysfunction. Autophagy is a remarkable patho (physiological) process in the cell homeostasis regulation including EC. Regulation of autophagy rate is disease-dependent and impaired with aging. Up-regulation of autophagy induces endothelial cell regeneration/differentiation and improves the function of impaired ones. The paper scrutinizes the molecular mechanisms and triggers of EC dysregulation and current perspectives for future therapeutic strategies by autophagy targeting.
Collapse
|
12
|
Sellahewa SG, Li JY, Xiao Q. Updated Perspectives on Direct Vascular Cellular Reprogramming and Their Potential Applications in Tissue Engineered Vascular Grafts. J Funct Biomater 2022; 14:21. [PMID: 36662068 PMCID: PMC9866165 DOI: 10.3390/jfb14010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular disease is a globally prevalent disease with far-reaching medical and socio-economic consequences. Although improvements in treatment pathways and revascularisation therapies have slowed disease progression, contemporary management fails to modulate the underlying atherosclerotic process and sustainably replace damaged arterial tissue. Direct cellular reprogramming is a rapidly evolving and innovative tissue regenerative approach that holds promise to restore functional vasculature and restore blood perfusion. The approach utilises cell plasticity to directly convert somatic cells to another cell fate without a pluripotent stage. In this narrative literature review, we comprehensively analyse and compare direct reprogramming protocols to generate endothelial cells, vascular smooth muscle cells and vascular progenitors. Specifically, we carefully examine the reprogramming factors, their molecular mechanisms, conversion efficacies and therapeutic benefits for each induced vascular cell. Attention is given to the application of these novel approaches with tissue engineered vascular grafts as a therapeutic and disease-modelling platform for cardiovascular diseases. We conclude with a discussion on the ethics of direct reprogramming, its current challenges, and future perspectives.
Collapse
Affiliation(s)
- Saneth Gavishka Sellahewa
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jojo Yijiao Li
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Qingzhong Xiao
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
13
|
Jung C, Oh JE, Lee S, Yoon YS. Generation and Application of Directly Reprogrammed Endothelial Cells. Korean Circ J 2022; 52:643-658. [PMID: 36097834 PMCID: PMC9470489 DOI: 10.4070/kcj.2022.0190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/15/2022] Open
Abstract
Cell-based therapy has emerged as a promising option for treating advanced ischemic cardiovascular disease by inducing vascular regeneration. However, clinical trials with adult cells turned out disappointing in general. As a newer approach, direct reprogramming has emerged to efficiently generate endothelial cells (ECs), which can promote neovascularization and vascular regeneration. This review provides recent updates on the direct endothelial reprogramming. In general, directly reprogrammed ECs can be generated by two approaches: one by transitioning through a plastic intermediate state and the other in a one-step transition without any intermediate states toward pluripotency. Moreover, the methods to deliver reprogramming factors and chemicals for the fate conversion are highlighted. Next, the therapeutic effects of the directly reprogrammed ECs on animal models are reviewed in detail. Other applications using directly reprogrammed ECs, such as tissue engineering and disease modeling, are also discussed. Lastly, the remaining questions and foremost challenges are addressed.
Collapse
Affiliation(s)
- Cholomi Jung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Eun Oh
- Research and Development Center, KarisBio Inc., Seoul, Korea
| | - Sangho Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Young-Sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Research and Development Center, KarisBio Inc., Seoul, Korea
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Mylonas KS, Karangelis D, Androutsopoulou V, Chalikias G, Tziakas D, Mikroulis D, Iliopoulos DC, Nikiteas N, Schizas D. Stem cell genes in atheromatosis: The role of
Klotho
,
HIF1α
,
OCT4
, and
BMP4. IUBMB Life 2022; 74:1003-1011. [DOI: 10.1002/iub.2667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023]
Affiliation(s)
- Konstantinos S. Mylonas
- Department of Cardiac Surgery Onassis Cardiac Surgery Center Athens Greece
- Laboratory of Experimental Surgery and Surgical Research “N.S. Christeas”, School of Medicine National and Kapodistrian University of Athens Athens Greece
| | - Dimos Karangelis
- Department of Cardiac Surgery, Democritus University of Thrace University Hospital of Alexandroupolis Alexandroupolis Greece
| | - Vasiliki Androutsopoulou
- Department of Cardiac Surgery, Democritus University of Thrace University Hospital of Alexandroupolis Alexandroupolis Greece
| | - George Chalikias
- Cardiology Department, Democritus University of Thrace University Hospital of Alexandroupolis Alexandroupolis Greece
| | - Dimitrios Tziakas
- Cardiology Department, Democritus University of Thrace University Hospital of Alexandroupolis Alexandroupolis Greece
| | - Dimitrios Mikroulis
- Department of Cardiac Surgery, Democritus University of Thrace University Hospital of Alexandroupolis Alexandroupolis Greece
| | - Dimitrios C. Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research “N.S. Christeas”, School of Medicine National and Kapodistrian University of Athens Athens Greece
- Fourth Department of Cardiac Surgery HYGEIA Hospital Athens Greece
| | - Nikolaos Nikiteas
- Second Propaedeutic Department of Surgery, Laiko General Hospital, School of Medicine National and Kapodistrian University of Athens Athens Greece
| | - Dimitrios Schizas
- First Department of Surgery, Laiko General Hospital, School of Medicine National and Kapodistrian University of Athens Athens Greece
| |
Collapse
|
15
|
Human Induced Pluripotent Stem Cell-Derived Vascular Cells: Recent Progress and Future Directions. J Cardiovasc Dev Dis 2021; 8:jcdd8110148. [PMID: 34821701 PMCID: PMC8622843 DOI: 10.3390/jcdd8110148] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great promise for cardiovascular regeneration following ischemic injury. Considerable effort has been made toward the development and optimization of methods to differentiate hiPSCs into vascular cells, such as endothelial and smooth muscle cells (ECs and SMCs). In particular, hiPSC-derived ECs have shown robust potential for promoting neovascularization in animal models of cardiovascular diseases, potentially achieving significant and sustained therapeutic benefits. However, the use of hiPSC-derived SMCs that possess high therapeutic relevance is a relatively new area of investigation, still in the earlier investigational stages. In this review, we first discuss different methodologies to derive vascular cells from hiPSCs with a particular emphasis on the role of key developmental signals. Furthermore, we propose a standardized framework for assessing and defining the EC and SMC identity that might be suitable for inducing tissue repair and regeneration. We then highlight the regenerative effects of hiPSC-derived vascular cells on animal models of myocardial infarction and hindlimb ischemia. Finally, we address several obstacles that need to be overcome to fully implement the use of hiPSC-derived vascular cells for clinical application.
Collapse
|
16
|
Wingo M, Rafii S. Endothelial reprogramming for vascular regeneration: Past milestones and future directions. Semin Cell Dev Biol 2021; 122:50-55. [PMID: 34548212 DOI: 10.1016/j.semcdb.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Endothelial cells are critical mediators of health and disease. Regenerative medicine techniques that target the endothelium hold vast promise for improving lifespan and quality of life worldwide. Regenerative therapies via induced pluripotent stem cells (IPSCs) have helped demonstrate disease mechanisms, but so far, concerns regarding their function, malignant potential, and expense have limited therapeutic potential. One alternative approach is direct reprogramming of somatic cells, which avoids the pluripotent state and allows for in vivo reprogramming. Transcription factors from endothelial development have yielded essential transcription factors and small molecules that induce endothelial cell fate. Most direct cell reprogramming strategies targeting endothelial cells use ETV2, a pioneer transcription factor to specify endothelial lineage via histone-modifying enzymes. Many different types of starting cells and strategies, including lentiviral transduction, inducing innate immunity, and small molecule signaling have been leveraged for reprogramming. However, so far therapeutic benefit of these strategies remains unproven. Future research will have to solve scalability, safety, and efficacy hurdles before being ready for the clinic. However, researchers have already discovered meaningful insights into disease mechanisms and development through direct reprogramming.
Collapse
Affiliation(s)
- Matthew Wingo
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
17
|
Hywood JD, Sadeghipour S, Clayton ZE, Yuan J, Stubbs C, Wong JWT, Cooke JP, Patel S. Induced endothelial cells from peripheral arterial disease patients and neonatal fibroblasts have comparable angiogenic properties. PLoS One 2021; 16:e0255075. [PMID: 34375370 PMCID: PMC8354451 DOI: 10.1371/journal.pone.0255075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/11/2021] [Indexed: 12/05/2022] Open
Abstract
Induced endothelial cells (iECs) generated from neonatal fibroblasts via transdifferentiation have been shown to have pro-angiogenic properties and are a potential therapy for peripheral arterial disease (PAD). It is unknown if iECs can be generated from fibroblasts collected from PAD patients and whether these cells are pro-angiogenic. In this study fibroblasts were collected from four PAD patients undergoing carotid endarterectomies. These cells, and neonatal fibroblasts, were transdifferentiated into iECs using modified mRNA. Endothelial phenotype and pro-angiogenic cytokine secretion were investigated. NOD-SCID mice underwent surgery to induce hindlimb ischaemia in a murine model of PAD. Mice received intramuscular injections with either control vehicle, or 1 × 106 neonatal-derived or 1 × 106 patient-derived iECs. Recovery in perfusion to the affected limb was measured using laser Doppler scanning. Perfusion recovery was enhanced in mice treated with neonatal-derived iECs and in two of the three patient-derived iEC lines investigated in vivo. Patient-derived iECs can be successfully generated from PAD patients and for specific patients display comparable pro-angiogenic properties to neonatal-derived iECs.
Collapse
Affiliation(s)
- Jack D. Hywood
- Heart Research Institute, Newtown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | | | - Zoe E. Clayton
- Heart Research Institute, Newtown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Jun Yuan
- Heart Research Institute, Newtown, NSW, Australia
| | - Colleen Stubbs
- RNACore, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Jack W. T. Wong
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Sanjay Patel
- Heart Research Institute, Newtown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
18
|
Zhou W, Ma T, Ding S. Non-viral approaches for somatic cell reprogramming into cardiomyocytes. Semin Cell Dev Biol 2021; 122:28-36. [PMID: 34238675 DOI: 10.1016/j.semcdb.2021.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/04/2021] [Accepted: 06/23/2021] [Indexed: 11/27/2022]
Abstract
Heart disease is the leading cause of human deaths worldwide. Due to lacking cardiomyocytes with replicative capacity and cardiac progenitor cells with differentiation potential in adult hearts, massive loss of cardiomyocytes after ischemic events produces permanent damage, ultimately leading to heart failure. Cellular reprogramming is a promising strategy to regenerate heart by induction of cardiomyocytes from other cell types, such as cardiac fibroblasts. In contrast to conventional virus-based cardiac reprogramming, non-viral approaches greatly reduce the potential risk that includes disruption of genome integrity by integration of foreign DNAs, expression of exogenous genes with oncogenic potential, and appearance of partially reprogrammed cells harmful for the physiological functions of tissues/organs, which impedes their in-vivo applications. Here, we review the recent progress in development of non-viral approaches to directly reprogram somatic cells towards cardiomyocytes and their therapeutic application for heart regeneration.
Collapse
Affiliation(s)
- Wei Zhou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Tianhua Ma
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Sheng Ding
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
19
|
Yi B, Ding T, Jiang S, Gong T, Chopra H, Sha O, Dissanayaka WL, Ge S, Zhang C. Conversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors. Stem Cell Res Ther 2021; 12:266. [PMID: 33941255 PMCID: PMC8091697 DOI: 10.1186/s13287-021-02350-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Objectives Recently, a new strategy has been developed to directly reprogram one cell type towards another targeted cell type using small molecule compounds. Human fibroblasts have been chemically reprogrammed into neuronal cells, Schwann cells and cardiomyocyte-like cells by different small molecule combinations. This study aimed to explore whether stem cells from apical papilla (SCAP) could be reprogrammed into endothelial cells (ECs) using the same strategy. Materials and methods The expression level of endothelial-specific genes and proteins after chemical induction of SCAP was assessed by RT-PCR, western blotting, flow cytometry and immunofluorescence. The in vitro functions of SCAP-derived chemical-induced endothelial cells (SCAP-ECs) were evaluated by tube-like structure formation assay, acetylated low-density lipoprotein (ac-LDL) uptake and NO secretion detection. The proliferation and the migration ability of SCAP-ECs were evaluated by CCK-8 and Transwell assay. LPS stimulation was used to mimic the inflammatory environment in demonstrating the ability of SCAP-ECs to express adhesion molecules. The in vivo Matrigel plug angiogenesis assay was performed to assess the function of SCAP-ECs in generating vascular structures using the immune-deficient mouse model. Results SCAP-ECs expressed upregulated endothelial-specific genes and proteins; displayed endothelial transcriptional networks; exhibited the ability to form functional tubular-like structures, uptake ac-LDL and secrete NO in vitro; and contributed to generate blood vessels in vivo. The SCAP-ECs could also express adhesion molecules in the pro-inflammatory environment and have a similar migration and proliferation ability as HUVECs. Conclusions Our study demonstrates that the set of small molecules and growth factors could significantly promote endothelial transdifferentiation of SCAP, which provides a promising candidate cell source for vascular engineering and treatment of ischemic diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02350-5.
Collapse
Affiliation(s)
- Baicheng Yi
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Tian Ding
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, Shandong, China
| | - Shan Jiang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ting Gong
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hitesh Chopra
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ou Sha
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
| | - Waruna Lakmal Dissanayaka
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Shaohua Ge
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University; Shandong Key Laboratory of Oral Tissue Regeneration; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No.44-1 Wenhua Road West, Jinan, Shandong, China.
| | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region, China. .,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China.
| |
Collapse
|
20
|
Liu HL, Tang HT, Yang HL, Deng TT, Xu YP, Xu SQ, Peng L, Wang Z, Fang Q, Kuang XY, Li QS. Oct4 Regulates the Transition of Cancer Stem-Like Cells to Tumor Endothelial-Like Cells in Human Liver Cancer. Front Cell Dev Biol 2020; 8:563316. [PMID: 33102474 PMCID: PMC7554317 DOI: 10.3389/fcell.2020.563316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Octamer-binding transcription factor 4 (Oct4) has been recently implicated as a proangiogenic regulator in several induced pluripotent stem cells (iPSCs), however, its role in cancer stem-like cells (CSCs) remain unclear. We report here that Oct4 participates in tumor vasculogenesis in liver CSCs (LCSCs). We identify that LCSCs possess the potential of endothelial trans-differentiation under endothelial induction, present endothelial specific markers and their functions in vitro, and participate in neovasculogenesis in vivo. The knockdown of the Oct4A by short hairpin RNA (shRNA) in LCSCs represses endothelial trans-differentiation potential, but induces endothelial lineage-restricted differentiation, the latter is positively regulated by Oct4B1. Furthermore, Oct4 regulates vasculogenesis in LCSCs may be via the AKT-NF-κB-p65 signaling pathway. This work reveals Oct4, which is a crucial regulator, plays a critical role in tumor endothelial-like cells transition of LCSCs through Oct4A and Oct4B1 by different ways. The simultaneous inhibition of both the isoforms of Oct4 is hence expected to help regress neovascularization derived from CSCs. Our findings may provide insights to the possible new mechanisms of tumor vasculogenesis for primary liver cancer.
Collapse
Affiliation(s)
- Hong-Lin Liu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Hong-Ting Tang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Han-Lin Yang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Ting-Ting Deng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Ya-Ping Xu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Shi-Qing Xu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Liang Peng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Qing Fang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiao-Yan Kuang
- Institute of Clinical Pathology, Zunyi Medical College, Zunyi, China
| | - Qin-Shan Li
- Department of Obstetrics and Gynecology, Prenatal Diagnosis Center, Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| |
Collapse
|
21
|
Yi B, Dissanayaka WL, Zhang C. Growth Factors and Small-molecule Compounds in Derivation of Endothelial Lineages from Dental Stem Cells. J Endod 2020; 46:S63-S70. [PMID: 32950197 DOI: 10.1016/j.joen.2020.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Incorporating fully assembled microvascular networks into bioengineered dental pulp constructs can significantly enhance functional blood flow and tissue survival upon transplantation. Endothelial cells (ECs), cellular building blocks of vascular tissue, play an essential role in the process of prevascularization. However, obtaining sufficient ECs from a suitable source for translational application is challenging. Dental stem cells (DSCs), which exhibit a robust proliferative ability and immunocompatibility because of their autologous origin, could be a promising alternative cell source for the derivation of endothelial lineages. Under specific culture conditions, DSCs differentiate into osteo/odontogenic, adipogenic, chondrogenic, and neurogenic cell lineages. METHODS Recently, a new approach has been developed to directly reprogram cells using chemical cocktails and growth factors. Compared with the traditional reprogramming approach based on the forced expression of exogenous transcription factors, the chemical strategy avoids the risk associated with lentiviral transduction while offering a more viable methodology to drive cell lineage switch. The aim of this review was to unveil the concept of the use of small-molecule compounds and growth factors modulating key signaling pathways to derive ECs from DSCs. RESULTS In addition, our preliminary study showed that stem cells from the apical papilla could be induced into EC-like cells using small-molecule compounds and growth factors. These EC-like cells expressed endothelial specific genes (CD31 and VEGFR2) and proteins (CD31, VEGF receptor 2, and vascular endothelial cadherin) as well as gave rise to vessel-like tubular structures in vitro. CONCLUSIONS Our preliminary results suggest that chemical reprogramming might offer a novel way to generate EC-like cells from dental stem cells.
Collapse
Affiliation(s)
- Baicheng Yi
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Waruna Lakmal Dissanayaka
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Special Administrative Region, China.
| |
Collapse
|
22
|
Nazarnezhad S, Baino F, Kim HW, Webster TJ, Kargozar S. Electrospun Nanofibers for Improved Angiogenesis: Promises for Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1609. [PMID: 32824491 PMCID: PMC7466668 DOI: 10.3390/nano10081609] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/27/2022]
Abstract
Angiogenesis (or the development of new blood vessels) is a key event in tissue engineering and regenerative medicine; thus, a number of biomaterials have been developed and combined with stem cells and/or bioactive molecules to produce three-dimensional (3D) pro-angiogenic constructs. Among the various biomaterials, electrospun nanofibrous scaffolds offer great opportunities for pro-angiogenic approaches in tissue repair and regeneration. Nanofibers made of natural and synthetic polymers are often used to incorporate bioactive components (e.g., bioactive glasses (BGs)) and load biomolecules (e.g., vascular endothelial growth factor (VEGF)) that exert pro-angiogenic activity. Furthermore, seeding of specific types of stem cells (e.g., endothelial progenitor cells) onto nanofibrous scaffolds is considered as a valuable alternative for inducing angiogenesis. The effectiveness of these strategies has been extensively examined both in vitro and in vivo and the outcomes have shown promise in the reconstruction of hard and soft tissues (mainly bone and skin, respectively). However, the translational of electrospun scaffolds with pro-angiogenic molecules or cells is only at its beginning, requiring more research to prove their usefulness in the repair and regeneration of other highly-vascularized vital tissues and organs. This review will cover the latest progress in designing and developing pro-angiogenic electrospun nanofibers and evaluate their usefulness in a tissue engineering and regenerative medicine setting.
Collapse
Affiliation(s)
- Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran;
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Hae-Won Kim
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Korea;
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan 31116, Korea
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA;
| | - Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran;
| |
Collapse
|
23
|
Kim HS, Kim JY, Song CL, Jeong JE, Cho YS. Directly induced human Schwann cell precursors as a valuable source of Schwann cells. Stem Cell Res Ther 2020; 11:257. [PMID: 32586386 PMCID: PMC7318441 DOI: 10.1186/s13287-020-01772-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/08/2020] [Accepted: 06/14/2020] [Indexed: 12/13/2022] Open
Abstract
Background Schwann cells (SCs) are primarily responsible for regeneration and repair of the peripheral nervous system (PNS). Renewable and lineage-restricted SC precursors (SCPs) are considered highly desirable and promising cell sources for the production of SCs and for studies of SC lineage development, but SCPs are extremely limited. Here, we present a novel direct conversion strategy for the generation of human SCPs, capable of differentiating into functional SCs. Methods Easily accessible human skin fibroblast cells were directly induced into integration-free SCPs using episomal vectors (Oct3/4, Klf4, Sox2, L-Myc, Lin28 and p53 shRNA) under SCP lineage-specific chemically defined medium conditions. Induced SCPs (iSCPs) were further examined for their ability to differentiate into SCs. The identification and functionality of iSCPs and iSCP-differentiated SCs (iSCs) were confirmed according to morphology, lineage-specific markers, neurotropic factor secretion, and/or standard functional assays. Results Highly pure, Sox 10-positive of iSCPs (more than 95% purity) were generated from human skin fibroblasts within 3 weeks. Established iSCPs could be propagated in vitro while maintaining their SCP identity. Within 1 week, iSCPs could efficiently differentiate into SCs (more than 95% purity). The iSCs were capable of secreting various neurotrophic factors such as GDNF, NGF, BDNF, and NT-3. The in vitro myelinogenic potential of iSCs was assessed by myelinating cocultures using mouse dorsal root ganglion (DRG) neurons or human induced pluripotent stem cell (iPSC)-derived sensory neurons (HSNs). Furthermore, iSC transplantation promoted sciatic nerve repair and improved behavioral recovery in a mouse model of sciatic nerve crush injury in vivo. Conclusions We report a robust method for the generation of human iSCPs/iSCs that might serve as a promising cellular source for various regenerative biomedical research and applications, such as cell therapy and drug discovery, especially for the treatment of PNS injury and disorders.
Collapse
Affiliation(s)
- Han-Seop Kim
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Jae Yun Kim
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea.,Department of Bioscience, KRIBB School, University of Science & Technology, 113 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea
| | - Cho Lok Song
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea.,Department of Bioscience, KRIBB School, University of Science & Technology, 113 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea
| | - Ji Eun Jeong
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Yee Sook Cho
- Stem Cell Research Laboratory (SCRL), Immunotherapy Research Center (IRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea. .,Department of Bioscience, KRIBB School, University of Science & Technology, 113 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea.
| |
Collapse
|
24
|
Thakur G, Lee HJ, Jeon RH, Lee SL, Rho GJ. Small Molecule-Induced Pancreatic β-Like Cell Development: Mechanistic Approaches and Available Strategies. Int J Mol Sci 2020; 21:E2388. [PMID: 32235681 PMCID: PMC7178115 DOI: 10.3390/ijms21072388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a metabolic disease which affects not only glucose metabolism but also lipid and protein metabolism. It encompasses two major types: type 1 and 2 diabetes. Despite the different etiologies of type 1 and 2 diabetes mellitus (T1DM and T2DM, respectively), the defining features of the two forms are insulin deficiency and resistance, respectively. Stem cell therapy is an efficient method for the treatment of diabetes, which can be achieved by differentiating pancreatic β-like cells. The consistent generation of glucose-responsive insulin releasing cells remains challenging. In this review article, we present basic concepts of pancreatic organogenesis, which intermittently provides a basis for engineering differentiation procedures, mainly based on the use of small molecules. Small molecules are more auspicious than any other growth factors, as they have unique, valuable properties like cell-permeability, as well as a nonimmunogenic nature; furthermore, they offer immense benefits in terms of generating efficient functional beta-like cells. We also summarize advances in the generation of stem cell-derived pancreatic cell lineages, especially endocrine β-like cells or islet organoids. The successful induction of stem cells depends on the quantity and quality of available stem cells and the efficient use of small molecules.
Collapse
Affiliation(s)
- Gitika Thakur
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Hyeon-Jeong Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Ryoung-Hoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sung-Lim Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| |
Collapse
|
25
|
Chen T, Karamariti E, Hong X, Deng J, Wu Y, Gu W, Simpson R, Wong MM, Yu B, Hu Y, Qu A, Xu Q, Zhang L. DKK3 (Dikkopf-3) Transdifferentiates Fibroblasts Into Functional Endothelial Cells-Brief Report. Arterioscler Thromb Vasc Biol 2020; 39:765-773. [PMID: 30816803 DOI: 10.1161/atvbaha.118.311919] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective- To determine the role of a cytokine-like protein DKK3 (dikkopf-3) in directly transdifferentiating fibroblasts into endothelial cells (ECs) and the underlying mechanisms. Approach and Results- DKK3 overexpression in human fibroblasts under defined conditions for 4 days led to a notable change in cell morphology and progenitor gene expression. It was revealed that these cells went through mesenchymal-to-epithelial transition and subsequently expressed KDR (kinase insert domain receptor) at high levels. Further culture in EC defined media led to differentiation of these progenitors into functional ECs capable of angiogenesis both in vitro and in vivo, which was regulated by the VEGF (vascular endothelial growth factor)/miR (microRNA)-125a-5p/Stat3 (signal transducer and activator of transcription factor 3) axis. More importantly, fibroblast-derived ECs showed the ability to form a patent endothelium-like monolayer in tissue-engineered vascular grafts ex vivo. Conclusions- These data demonstrate that DKK3 is capable of directly differentiating human fibroblasts to functional ECs under defined media and provides a novel potential strategy for endothelial regeneration.
Collapse
Affiliation(s)
- Ting Chen
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (T.C., Y.W., Q.X., L.Z.)
| | - Eirini Karamariti
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Xuechong Hong
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Jiacheng Deng
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Yutao Wu
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (T.C., Y.W., Q.X., L.Z.)
| | - Wenduo Gu
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Russell Simpson
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Mei Mei Wong
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (B.Y., A.Q.)
| | - Yanhua Hu
- School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (B.Y., A.Q.)
| | - Qingbo Xu
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (T.C., Y.W., Q.X., L.Z.).,School of Cardiovascular Medicine and Sciences, King's College London BHF Centre, London, United Kingdom (E.K., X.H., J.D., W.D., R.S., M.M.W., Y.H., Q.X.)
| | - Li Zhang
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (T.C., Y.W., Q.X., L.Z.)
| |
Collapse
|
26
|
Hou L, Yang G, Tang S, Alcazar C, Joshi P, Strassberg Z, Kim M, Kawamura M, Woo YJ, Shrager J, Ding S, Huang NF. Small Molecule Derived From Carboxyethylpyrrole Protein Adducts Promotes Angiogenesis in a Mouse Model of Peripheral Arterial Disease. J Am Heart Assoc 2019; 7:e009234. [PMID: 30371212 PMCID: PMC6222956 DOI: 10.1161/jaha.118.009234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background CEP (ω-[2-carboxyethyl]pyrrole) protein adducts are the end products of lipid oxidation associated with inflammation and have been implicated in the induction of angiogenesis in pathological conditions such as tissue ischemia. We synthesized small molecules derived from CEP protein adducts and evaluated the angiogenic effect of the CEP analog CEP 03 in the setting of peripheral arterial disease. Methods and Results The angiogenic effect of CEP 03 was assessed by in vitro analysis of primary human microvascular endothelial cell proliferation and tubelike formation in Matrigel (Corning). In the presence of CEP 03, proliferation of endothelial cells in vitro increased by 27±18% under hypoxic (1% O2) conditions, reaching similar levels to that of VEGF A (vascular endothelial growth factor A) stimulation (22±10%), relative to the vehicle control treatment. A similar effect of CEP 03 was demonstrated in the increased number of tubelike branches in Matrigel, reaching >70% induction in hypoxia, compared with the vehicle control. The therapeutic potential of CEP 03 was further evaluated in a mouse model of peripheral arterial disease by quantification of blood perfusion recovery and capillary density. In the ischemic hind limb, treatment of CEP 03 encapsulated within Matrigel significantly enhanced blood perfusion by 2-fold after 14 days compared with those treated with Matrigel alone. Moreover, these results concurred with histological finding that treatment of CEP 03 in Matrigel resulted in a significant increase in microvessel density compared with Matrigel alone. Conclusions Our data suggest that CEP 03 has a profound positive effect on angiogenesis and neovessel formation and thus has therapeutic potential for treatment of peripheral arterial disease.
Collapse
Affiliation(s)
- Luqia Hou
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Guang Yang
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Shibing Tang
- 3 Gladstone Institute of Cardiovascular Disease University of California San Francisco CA
| | - Cynthia Alcazar
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Prajakta Joshi
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Zachary Strassberg
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Michael Kim
- 2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Masashi Kawamura
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Y Joseph Woo
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Joseph Shrager
- 2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Sheng Ding
- 3 Gladstone Institute of Cardiovascular Disease University of California San Francisco CA.,5 Department of Pharmaceutical Chemistry University of California San Francisco CA
| | - Ngan F Huang
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| |
Collapse
|
27
|
Liu Y, Zhao S, Luo L, Wang J, Zhu Z, Xiang Q, Deng Y, Zhao Z. Mesenchymal stem cell-derived exosomes ameliorate erection by reducing oxidative stress damage of corpus cavernosum in a rat model of artery injury. J Cell Mol Med 2019; 23:7462-7473. [PMID: 31512385 PMCID: PMC6815831 DOI: 10.1111/jcmm.14615] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/01/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
Erectile dysfunction (ED) is a common ageing male's disease, and vascular ED accounts for the largest proportion of all types of ED. One of the mechanisms of vascular ED in the clinic is arterial insufficiency, which mainly caused by atherosclerosis, trauma and surgical. Moreover, oxidative stress damage after tissue ischemia usually aggravated the progress of ED. As a new way of acellular therapy, mesenchymal stem cell-derived exosomes (MSC-Exos) have great potential in ED treatment. In the current study, we have explored the mechanism of MSC-Exos therapy in a rat model of internal iliac artery injury-induced ED. Compared with intracavernous (IC) injection of phosphate-buffered saline after artery injury, of note, we observed that both mesenchymal stem cells (MSCs) and MSC-Exos through IC injection could improve the erectile function to varying degrees. More specifically, IC injection MSC-Exos could promote cavernous sinus endothelial formation, reduce the organization oxidative stress damage, and improve the nitric oxide synthase and smooth muscle content in the corpus cavernosum. With similar potency compared with the stem cell therapy and other unique advantages, IC injection of MSC- Exos could be an effective treatment to ameliorate erectile function in a rat model of arterial injury.
Collapse
Affiliation(s)
- Yangzhou Liu
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shankun Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Urology, Zhejiang Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Lianmin Luo
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiamin Wang
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiguo Zhu
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Xiang
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yihan Deng
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhigang Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
28
|
Wong AP, Shojaie S, Liang Q, Xia S, Di Paola M, Ahmadi S, Bilodeau C, Garner J, Post M, Duchesneau P, Waddell TK, Bear CE, Nagy A, Rossant J. Conversion of human and mouse fibroblasts into lung-like epithelial cells. Sci Rep 2019; 9:9027. [PMID: 31227724 PMCID: PMC6588580 DOI: 10.1038/s41598-019-45195-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/31/2019] [Indexed: 01/08/2023] Open
Abstract
Cell lineage conversion of fibroblasts to specialized cell types through transdifferentiation may provide a fast and alternative cell source for regenerative medicine. Here we show that transient transduction of fibroblasts with the four reprogramming factors (Oct4, Sox2, Klf4, and c-Myc) in addition to the early lung transcription factor Nkx2-1 (also known as Ttf1), followed by directed differentiation of the cells, can convert mouse embryonic and human adult dermal fibroblasts into induced lung-like epithelial cells (iLEC). These iLEC differentiate into multiple lung cell types in air liquid interface cultures, repopulate decellularized rat lung scaffolds, and form lung epithelia composed of Ciliated, Goblet, Basal, and Club cells after transplantation into immune-compromised mice. As proof-of-concept, differentiated human iLEC harboring the Cystic Fibrosis mutation dF508 demonstrated pharmacological rescue of CFTR function using the combination of lumacaftor and ivacaftor. Overall, this is a promising alternative approach for generation of patient-specific lung-like progenitors to study lung function, disease and future regeneration strategies.
Collapse
Affiliation(s)
- Amy P Wong
- Program in Developmental & Stem Cell Biology, SickKids Research Institute, Hospital for Sick Children, Toronto, ON, Canada.
| | - Sharareh Shojaie
- Program in Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Qin Liang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Sunny Xia
- Program in Molecular Medicine, SickKids Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Michelle Di Paola
- Program in Molecular Medicine, SickKids Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Saumel Ahmadi
- Program in Molecular Medicine, SickKids Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Claudia Bilodeau
- Program in Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Jodi Garner
- Program in Developmental & Stem Cell Biology, SickKids Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Martin Post
- Program in Translational Medicine, SickKids Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Pascal Duchesneau
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, and the McEwen Centre for Regenerative Medicine, Toronto, Canada
| | - Thomas K Waddell
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, and the McEwen Centre for Regenerative Medicine, Toronto, Canada
| | - Christine E Bear
- Program in Molecular Medicine, SickKids Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Janet Rossant
- Program in Developmental & Stem Cell Biology, SickKids Research Institute, Hospital for Sick Children, Toronto, ON, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada. .,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Canada.
| |
Collapse
|
29
|
Talkhabi M. Partial reprogramming as a therapeutic approach for heart disease: A state-of-the-art review. J Cell Biochem 2019; 120:14247-14261. [PMID: 31081174 DOI: 10.1002/jcb.28900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/10/2019] [Accepted: 03/22/2019] [Indexed: 11/08/2022]
Abstract
Heart disease such as myocardial infarction is the first cause of mortality in all countries. Today, cardiac cell-based therapy using de novo produced cardiac cells is considered as a novel approach for cardiac regenerative medicine. Recently, an alchemy-like approach, known as direct reprogramming or direct conversion, has been developed to directly convert somatic cells to cardiac cells in vitro and in vivo. This cellular alchemy is a short-cut and safe strategy for generating autologous cardiac cells, and it can be accomplished through activating cardiogenesis- or pluripotency-related factors in noncardiac cells. Importantly, pluripotency factors-based direct cardiac conversion, known as partial reprogramming, is shorter and more efficient for cardiomyocyte generation in vitro. Today, this strategy is achievable for direct conversion of mouse and human somatic cells to cardiac lineage cells (cardiomyocytes and cardiac progenitor cells), using transgene free, chemical-based approaches. Although, heart-specific partial reprogramming seems to be challenging for in vivo conversion of cardiac fibroblasts to cardiac cells, but whole organism-based in vivo partial reprogramming ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in mouse. Notably, cardiac cells produced using partial reprogramming strategy can be a useful platform for disease modeling, drug screening and cardiac cell-based therapy, once the safety issues are overcome. Herein, we discuss about all progresses in de novo production of cardiac cells using partial reprogramming-based direct conversion, as well as give an overview about the potential applications of this strategy in vivo and in vitro.
Collapse
Affiliation(s)
- Mahmood Talkhabi
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
30
|
Williams IM, Wu JC. Generation of Endothelial Cells From Human Pluripotent Stem Cells. Arterioscler Thromb Vasc Biol 2019; 39:1317-1329. [PMID: 31242035 DOI: 10.1161/atvbaha.119.312265] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endothelial cells (ECs) are critical for several aspects of cardiovascular disease therapy, including vascular regeneration, personalized drug development, and tissue engineering. Human pluripotent stem cells (hPSCs) afford us with an unprecedented opportunity to produce virtually unlimited quantities of human ECs. In this review, we highlight key developments and outstanding challenges in our ability to derive ECs de novo from hPSCs. Furthermore, we consider strategies for recapitulating the vessel- and tissue-specific functional heterogeneity of ECs in vitro. Finally, we discuss ongoing attempts to utilize hPSC-derived ECs and their progenitors for various therapeutic applications. Continued progress in generating hPSC-derived ECs will profoundly enhance our ability to discover novel drug targets, revascularize ischemic tissues, and engineer clinically relevant tissue constructs. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Ian M Williams
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA
| |
Collapse
|
31
|
Biofabrication of a vascularized islet organ for type 1 diabetes. Biomaterials 2019; 199:40-51. [PMID: 30735895 DOI: 10.1016/j.biomaterials.2019.01.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/27/2018] [Accepted: 01/18/2019] [Indexed: 12/17/2022]
Abstract
Islet transplantation is superior to extrinsic insulin supplementation in the treating severe Type 1 diabetes. However, its efficiency and longevity are limited by substantial islet loss post-transplantation due to lack of engraftment and vascular supply. To overcome these limitations, we developed a novel approach to bio-fabricate functional, vascularized islet organs (VIOs) ex vivo. We endothelialized acellular lung matrixes to provide a biocompatible multicompartment scaffold with an intact hierarchical vascular tree as a backbone for islet engraftment. Over seven days of culture, islets anatomically and functionally integrated into the surrounding bio-engineered vasculature, generating a functional perfusable endocrine organ. When exposed to supra-physiologic arterial glucose levels in vivo and ex vivo, mature VIOs responded with a physiologic insulin release from the vein and provided more efficient reduction of hyperglycemia compared to intraportally transplanted fresh islets. In long-term transplants in diabetic mice, subcutaneously implanted VIOs achieved normoglycemia significantly faster and more efficiently compared to islets that were transplanted in deviceless fashion. We conclude that ex vivo bio-fabrication of VIOs enables islet engraftment and vascularization before transplantation, and thereby helps to overcome limited islet survival and function observed in conventional islet transplantation. Given recent progress in stem cells, this technology may enable assembly of functional personalized endocrine organs.
Collapse
|
32
|
Zare L, Baharvand H, Javan M. In vivo conversion of astrocytes to oligodendrocyte lineage cells using chemicals: targeting gliosis for myelin repair. Regen Med 2018; 13:803-819. [PMID: 30284949 DOI: 10.2217/rme-2017-0155] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM It would be clinically ideal to target astrocytes in vivo for conversion into oligodendrocyte lineage cells to reduce astrogliosis and generate new myelinating cells. MATERIALS & METHODS Here, we prepared a GFP-labeled human astrocyte cell line, treated with epigenetic modifiers trichostatin A or 5-azacytidine and transplanted them into cuprizone-induced demyelinated mice brains. The fate of the transplanted astrocytes was studied at days 7, 14 and 28 post-transplantation. RESULTS GFP+ astrocytes were reduced over time, whereas the GFP+ oligodendrocyte lineage cells were found on days 14 and 28. Nontreated astrocytes did not convert to myelinating cells following transplantation. Cell conversion was proved in vitro by maintaining the treated cells in oligodendrocyte progenitor cell medium. CONCLUSION These findings seem promising for the application of epigenetic modifiers, especially their targeted delivery to glial scars to treat demyelinating diseases.
Collapse
Affiliation(s)
- Leila Zare
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran
| | - Hossein Baharvand
- Department of Brain Sciences & Cognition, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran 16635-148, Iran.,Department of Developmental Biology, University of Science & Culture, Tehran 1461968151, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran.,Department of Brain Sciences & Cognition, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran 16635-148, Iran
| |
Collapse
|
33
|
Tsifaki M, Kelaini S, Caines R, Yang C, Margariti A. Regenerating the Cardiovascular System Through Cell Reprogramming; Current Approaches and a Look Into the Future. Front Cardiovasc Med 2018; 5:109. [PMID: 30177971 PMCID: PMC6109758 DOI: 10.3389/fcvm.2018.00109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease (CVD), despite the advances of the medical field, remains one of the leading causes of mortality worldwide. Discovering novel treatments based on cell therapy or drugs is critical, and induced pluripotent stem cells (iPS Cells) technology has made it possible to design extensive disease-specific in vitro models. Elucidating the differentiation process challenged our previous knowledge of cell plasticity and capabilities and allows the concept of cell reprogramming technology to be established, which has inspired the creation of both in vitro and in vivo techniques. Patient-specific cell lines provide the opportunity of studying their pathophysiology in vitro, which can lead to novel drug development. At the same time, in vivo models have been designed where in situ transdifferentiation of cell populations into cardiomyocytes or endothelial cells (ECs) give hope toward effective cell therapies. Unfortunately, the efficiency as well as the concerns about the safety of all these methods make it exceedingly difficult to pass to the clinical trial phase. It is our opinion that creating an ex vivo model out of patient-specific cells will be one of the most important goals in the future to help surpass all these hindrances. Thus, in this review we aim to present the current state of research in reprogramming toward the cardiovascular system's regeneration, and showcase how the development and study of a multicellular 3D ex vivo model will improve our fighting chances.
Collapse
Affiliation(s)
- Marianna Tsifaki
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Sophia Kelaini
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
34
|
Meng S, Chanda P, Thandavarayan RA, Cooke JP. Transflammation: How Innate Immune Activation and Free Radicals Drive Nuclear Reprogramming. Antioxid Redox Signal 2018; 29:205-218. [PMID: 29634341 PMCID: PMC6003401 DOI: 10.1089/ars.2017.7364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Yamanaka and colleagues galvanized the field of stem cell biology and regenerative medicine by their generation of induced pluripotent stem cells. Evidence is emerging that activation of innate immune signaling is critical for efficient reprogramming to pluripotency and for the nuclear reprogramming occurring in transdifferentiation. Recent Advances: We have shown that innate immune signaling triggers a global change in the expression of epigenetic modifiers to enhance DNA accessibility. In this state of epigenetic plasticity, overexpression of lineage determination factors, and/or environmental cues and paracrine factors, can induce pluripotency, or can direct transdifferentiation to another somatic cell lineage. Accumulating evidence reveals that innate immune activation triggers the generation of reactive oxygen species and reactive nitrogen species, and that these free radicals are required for nuclear reprogramming to pluripotency or for transdifferentiation. CRITICAL ISSUES We have discovered a limb of innate immune signaling that regulates DNA accessibility, in part, by the action of free radicals to induce post-translational modification of epigenetic modifiers. FUTURE DIRECTIONS It is of scientific interest and clinical relevance to understand the mechanisms by which free radicals influence epigenetic plasticity, and how these mechanisms may be therapeutically modulated. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Shu Meng
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| | - Palas Chanda
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| | - Rajarajan A Thandavarayan
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| | - John P Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute , Houston, Texas
| |
Collapse
|
35
|
Malik V, Zimmer D, Jauch R. Diversity among POU transcription factors in chromatin recognition and cell fate reprogramming. Cell Mol Life Sci 2018; 75:1587-1612. [PMID: 29335749 PMCID: PMC11105716 DOI: 10.1007/s00018-018-2748-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/23/2017] [Accepted: 01/08/2018] [Indexed: 12/28/2022]
Abstract
The POU (Pit-Oct-Unc) protein family is an evolutionary ancient group of transcription factors (TFs) that bind specific DNA sequences to direct gene expression programs. The fundamental importance of POU TFs to orchestrate embryonic development and to direct cellular fate decisions is well established, but the molecular basis for this activity is insufficiently understood. POU TFs possess a bipartite 'two-in-one' DNA binding domain consisting of two independently folding structural units connected by a poorly conserved and flexible linker. Therefore, they represent a paradigmatic example to study the molecular basis for the functional versatility of TFs. Their modular architecture endows POU TFs with the capacity to accommodate alternative composite DNA sequences by adopting different quaternary structures. Moreover, associations with partner proteins crucially influence the selection of their DNA binding sites. The plentitude of DNA binding modes confers the ability to POU TFs to regulate distinct genes in the context of different cellular environments. Likewise, different binding modes of POU proteins to DNA could trigger alternative regulatory responses in the context of different genomic locations of the same cell. Prominent POU TFs such as Oct4, Brn2, Oct6 and Brn4 are not only essential regulators of development but have also been successfully employed to reprogram somatic cells to pluripotency and neural lineages. Here we review biochemical, structural, genomic and cellular reprogramming studies to examine how the ability of POU TFs to select regulatory DNA, alone or with partner factors, is tied to their capacity to epigenetically remodel chromatin and drive specific regulatory programs that give cells their identities.
Collapse
Affiliation(s)
- Vikas Malik
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- Genome Regulation Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Dennis Zimmer
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- Genome Regulation Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ralf Jauch
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
- Genome Regulation Laboratory, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
36
|
Tan RP, Chan AHP, Lennartsson K, Miravet MM, Lee BSL, Rnjak-Kovacina J, Clayton ZE, Cooke JP, Ng MKC, Patel S, Wise SG. Integration of induced pluripotent stem cell-derived endothelial cells with polycaprolactone/gelatin-based electrospun scaffolds for enhanced therapeutic angiogenesis. Stem Cell Res Ther 2018; 9:70. [PMID: 29562916 PMCID: PMC5863387 DOI: 10.1186/s13287-018-0824-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/19/2018] [Accepted: 03/05/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Induced pluripotent stem-cell derived endothelial cells (iPSC-ECs) can be generated from any somatic cell and their iPSC sources possess unlimited self-renewal. Previous demonstration of their proangiogenic activity makes them a promising cell type for treatment of ischemic injury. As with many other stem cell approaches, the low rate of in-vivo survival has been a major limitation to the efficacy of iPSC-ECs to date. In this study, we aimed to increase the in-vivo lifetime of iPSC-ECs by culturing them on electrospun polycaprolactone (PCL)/gelatin scaffolds, before quantifying the subsequent impact on their proangiogenic function. METHODS iPSC-ECs were isolated and stably transfected with a luciferase reporter to facilitate quantification of cell numbers and non-invasive imaging in-vivo PCL/gelatin scaffolds were engineered using electrospinning to obtain woven meshes of nanofibers. iPSC-ECs were cultured on scaffolds for 7 days. Subsequently, cell growth and function were assessed in vitro followed by implantation in a mouseback subcutaneous model for 7 days. RESULTS Using a matrix of conditions, we found that scaffold blends with ratios of PCL:gelatin of 70:30 (PG73) spun at high flow rates supported the greatest levels of iPSC-EC growth, retention of phenotype, and function in vitro. Implanting iPSC-ECs seeded on PG73 scaffolds in vivo improved their survival up to 3 days, compared to cells directly injected into control wounds, which were no longer observable within 1 h. Enhanced engraftment improved blood perfusion, observed through non-invasive laser Doppler imaging. Immunohistochemistry revealed a corresponding increase in host angiogenic mechanisms characterized by the enhanced recruitment of macrophages and the elevated expression of proangiogenic cytokines vascular endothelial growth factor and placental growth factor. CONCLUSIONS Knowledge of these mechanisms combined with a deeper understanding of the scaffold parameters influencing this function provides the groundwork for optimizing future iPSC-EC therapies utilizing engraftment platforms. The development of combined scaffold and iPSC-EC therapies could ultimately improve therapeutic angiogenesis and the treatment of ischemic injury.
Collapse
Affiliation(s)
- Richard P Tan
- The Heart Research Institute, Sydney, NSW, 2042, Australia. .,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Alex H P Chan
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | | | | | - Bob S L Lee
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zoe E Clayton
- The Heart Research Institute, Sydney, NSW, 2042, Australia
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Martin K C Ng
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Royal Prince Alfred Hospital, Sydney, NSW, 2042, Australia
| | - Sanjay Patel
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Royal Prince Alfred Hospital, Sydney, NSW, 2042, Australia
| | - Steven G Wise
- The Heart Research Institute, Sydney, NSW, 2042, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
37
|
FLI1 and PKC co-activation promote highly efficient differentiation of human embryonic stem cells into endothelial-like cells. Cell Death Dis 2018; 9:131. [PMID: 29374149 PMCID: PMC5833666 DOI: 10.1038/s41419-017-0162-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/24/2017] [Accepted: 11/13/2017] [Indexed: 12/28/2022]
Abstract
Rationale-endothelial cells (ECs) play important roles in various regeneration processes and can be used in a variety of therapeutic applications, such as cardiac regeneration, gene therapy, tissue-engineered vascular grafts and prevascularized tissue transplants. ECs can be acquired from pluripotent and adult stem cells. To acquire ECs from human embryonic stem cells (hESCs) in a fast, efficient and economic manner. We established a conditional overexpression system in hESCs based on 15 transcription factors reported to be responsible for hematopoiesis lineage. Among them, only overexpression of FLI1 could induce hESCs to a hematopoietic lineage. Moreover, simultaneous overexpression of FLI1 and activation of PKC rapidly and efficiently induced differentiation of hESCs into induced endothelial cells (iECs) within 3 days, while neither FLI1 overexpression nor PKC activation alone could derive iECs from hESCs. During induction, hESCs differentiated into spindle-like cells that were consistent in appearance with ECs. Flow cytometric analysis revealed that 92.2-98.9% and 87.2-92.6% of these cells were CD31+ and CD144+, respectively. Expression of vascular-specific genes dramatically increased, while the expression of pluripotency genes gradually decreased during induction. iECs incorporated acetylated low-density lipoproteins, strongly expressed vWF and bound UEA-1. iECs also formed capillary-like structures both in vitro and in vivo. RNA-seq analysis verified that these cells closely resembled their in vivo counterparts. Our results showed that co-activation of FLI1 and PKC could induce differentiation of hESCs into iECs in a fast, efficient and economic manner.
Collapse
|
38
|
Ghiroldi A, Piccoli M, Ciconte G, Pappone C, Anastasia L. Regenerating the human heart: direct reprogramming strategies and their current limitations. Basic Res Cardiol 2017; 112:68. [DOI: 10.1007/s00395-017-0655-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 10/12/2017] [Indexed: 12/15/2022]
|
39
|
Lin Y, Gil CH, Yoder MC. Differentiation, Evaluation, and Application of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Arterioscler Thromb Vasc Biol 2017; 37:2014-2025. [PMID: 29025705 DOI: 10.1161/atvbaha.117.309962] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The emergence of induced pluripotent stem cell (iPSC) technology paves the way to generate large numbers of patient-specific endothelial cells (ECs) that can be potentially delivered for regenerative medicine in patients with cardiovascular disease. In the last decade, numerous protocols that differentiate EC from iPSC have been developed by many groups. In this review, we will discuss several common strategies that have been optimized for human iPSC-EC differentiation and subsequent studies that have evaluated the potential of human iPSC-EC as a cell therapy or as a tool in disease modeling. In addition, we will emphasize the importance of using in vivo vessel-forming ability and in vitro clonogenic colony-forming potential as a gold standard with which to evaluate the quality of human iPSC-EC derived from various protocols.
Collapse
Affiliation(s)
- Yang Lin
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Chang-Hyun Gil
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Mervin C Yoder
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis.
| |
Collapse
|
40
|
|
41
|
Transflammation: Innate immune signaling in nuclear reprogramming. Adv Drug Deliv Rev 2017; 120:133-141. [PMID: 28916494 DOI: 10.1016/j.addr.2017.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/31/2017] [Accepted: 09/07/2017] [Indexed: 12/23/2022]
Abstract
Induction of pluripotency in somatic cells by retroviral overexpression of four transcription factors has revolutionized the field of stem cell biology and regenerative medicine. The efficient induction of pluripotency requires the activation of innate immune signaling in a process termed "transflammation" (Lee et al., 2012). Specifically, the stimulation of pattern recognition receptors (PRRs) causes global alterations in the expression and activity of epigenetic modifiers to favor an open chromatin configuration. Activation of toll-like receptors (TLR) or RIG-1-like receptors (RLR) (Sayed et al. 2017) trigger signaling cascades that result in NFκB or IRF-3 mediated changes in epigenetic plasticity that facilitate reprogramming. Another form of nuclear reprogramming is so-called direct reprogramming or transdifferentiation of one somatic cell to another lineage. We have shown that transdifferentiation of human fibroblasts to endothelial cells also involves transflammation (Sayed et al., 2015). Recently, we also identified reactive oxygen species (ROS) (Zhou et al. 2016) and reactive nitrogen species (RNS) (Meng et al., 2016) as mediators of innate immune signaling in nuclear reprogramming. Innate immune signaling plays a key role in nuclear reprogramming by regulating DNA accessibility (Fig. 1). Here, we review recent progress of innate immunity signaling in nuclear reprogramming and epigenetic plasticity.
Collapse
|
42
|
Akbari B, Wee P, Yaqubi M, Mohammadnia A. Comprehensive transcriptome mining of the direct conversion of mesodermal cells. Sci Rep 2017; 7:10427. [PMID: 28874788 PMCID: PMC5585404 DOI: 10.1038/s41598-017-10903-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/16/2017] [Indexed: 12/15/2022] Open
Abstract
The direct reprogramming of somatic cells is a promising approach for regenerative medicine, especially in the production of mesoderm layer-derived cells. Meta-analysis studies provide precise insight into the undergoing processes and help increase the efficiency of reprogramming. Here, using 27 high-throughput expression data sets, we analyzed the direct reprogramming of mesodermal cells in humans and mice. Fibroblast-derived cells showed a common expression pattern of up- and down-regulated genes that were mainly involved in the suppression of the fibroblast-specific gene expression program, and may be used as markers of the initiation of reprogramming. Furthermore, we found a specific gene expression profile for each fibroblast-derived cell studied, and each gene set appeared to play specific functional roles in its cell type, suggesting their use as markers for their mature state. Furthermore, using data from protein-DNA interactions, we identified the main transcription factors (TFs) involved in the conversion process and ranked them based on their importance in their gene regulatory networks. In summary, our meta-analysis approach provides new insights on the direct conversion of mesodermal somatic cells, introduces a list of genes as markers for initiation and maturation, and identifies TFs for which manipulating their expression may increase the efficiency of direct conversion.
Collapse
Affiliation(s)
- Bijan Akbari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ping Wee
- Department of Medical Genetics and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Moein Yaqubi
- Department of Psychiatry, Sackler Program for Epigenetics and Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
43
|
Mistriotis P, Andreadis ST. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res Rev 2017; 37:94-116. [PMID: 28579130 DOI: 10.1016/j.arr.2017.05.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Aging is the main risk factor contributing to vascular dysfunction and the progression of vascular diseases. In this review, we discuss the causes and mechanisms of vascular aging at the tissue and cellular level. We focus on Endothelial Cell (EC) and Smooth Muscle Cell (SMC) aging due to their critical role in mediating the defective vascular phenotype. We elaborate on two categories that contribute to cellular dysfunction: cell extrinsic and intrinsic factors. Extrinsic factors reflect systemic or environmental changes which alter EC and SMC homeostasis compromising vascular function. Intrinsic factors induce EC and SMC transformation resulting in cellular senescence. Replenishing or rejuvenating the aged/dysfunctional vascular cells is critical to the effective repair of the vasculature. As such, this review also elaborates on recent findings which indicate that stem cell and gene therapies may restore the impaired vascular cell function, reverse vascular aging, and prolong lifespan.
Collapse
Affiliation(s)
- Panagiotis Mistriotis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.
| |
Collapse
|
44
|
Transdifferentiated Human Vascular Smooth Muscle Cells are a New Potential Cell Source for Endothelial Regeneration. Sci Rep 2017; 7:5590. [PMID: 28717251 PMCID: PMC5514066 DOI: 10.1038/s41598-017-05665-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/01/2017] [Indexed: 01/04/2023] Open
Abstract
Endothelial dysfunction is widely implicated in cardiovascular pathological changes and development of vascular disease. In view of the fact that the spontaneous endothelial cell (EC) regeneration is a slow and insufficient process, it is of great interest to explore alternative cell sources capable of generating functional ECs. Vascular smooth muscle cell (SMC) composes the majority of the vascular wall and retains phenotypic plasticity in response to various stimuli. The aim of this study is to test the feasibility of the conversion of SMC into functional EC through the use of reprogramming factors. Human SMCs are first dedifferentiated for 4 days to achieve a vascular progenitor state expressing CD34, by introducing transcription factors OCT4, SOX2, KLF4 and c-MYC. These SMC-derived progenitors are then differentiated along the endothelial lineage. The SMC-converted ECs exhibit typical endothelial markers expression and endothelial functions in vitro, in vivo and in disease model. Further comprehensive analysis indicates that mesenchymal-to-epithelial transition is requisite to initiate SMCs reprogramming into vascular progenitors and that members of the Notch signalling pathway regulate further differentiation of the progenitors into endothelial lineage. Together, we provide the first evidence of the feasibility of the conversion of human SMCs towards endothelial lineage through an intermediate vascular progenitor state induced by reprogramming.
Collapse
|
45
|
Lee S, Kim JE, Johnson BA, Andukuri A, Yoon YS. Direct reprogramming into endothelial cells: a new source for vascular regeneration. Regen Med 2017. [PMID: 28621172 DOI: 10.2217/rme-2017-0022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Sangho Lee
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jin Eyun Kim
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brandon Al Johnson
- Wallace H Coulter Department of Biomedical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Adinarayana Andukuri
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Young-Sup Yoon
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Xie M, Tang S, Li K, Ding S. Pharmacological Reprogramming of Somatic Cells for Regenerative Medicine. Acc Chem Res 2017; 50:1202-1211. [PMID: 28453285 DOI: 10.1021/acs.accounts.7b00020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lost or damaged cells in tissues and organs can be replaced by transplanting therapeutically competent cells. This approach requires methods that effectively manipulate cellular identities and properties to generate sufficient numbers of desired cell types for transplantation. These cells can be generated by reprogramming readily available somatic cells, such as fibroblasts, into induced pluripotent stem cells (iPSCs), which can replicate indefinitely and give rise to any somatic cell type. This reprogramming can be achieved with genetic methods, such as forced expression of pluripotency-inducing transcription factors (TFs), which can be further improved, or even avoided, with pharmacological agents. We screened chemical libraries for such agents and identified small molecules that enhance TF-mediated pluripotency induction in somatic cells. We also developed cocktails of small molecules that can functionally replace combinations of TFs required to induce pluripotency in mouse and human somatic cells. Importantly, we devised and established a general strategy to develop effective pharmacological cocktails for specific cellular reprogramming processes. In the search for useful small molecules, we also discovered and characterized previously unknown mechanisms pertinent to cellular reprogramming. A more direct method to access scarce cells for cell transplantation is transdifferentiation, which uses combinations of cell-type specific TFs to reprogram fibroblasts into the target somatic cell types across lineage boundaries. We created an alternative strategy for cellular transdifferentiation that epigenetically activates somatic cells by pairing temporal treatment with reprogramming molecules and tissue-specific signaling molecules to generate cells of multiple lineages. Using this cell-activation and signaling-directed (CASD) transdifferentiation paradigm, we converted fibroblasts into a variety of somatic cells found in major organs, such as the heart, brain, pancreas, and liver. Specifically, we induced, isolated, and expanded (long-term) lineage-specific progenitor cells that can give rise to a defined range of cell types relevant to specific tissues or organs. Transplanting these progenitor cells or their progeny was therapeutically beneficial in animal models of diseases and organ damage. Importantly, we developed chemically defined conditions, without any genetic factors, that convert fibroblasts into cells of the cardiac and neural lineages, further extending the realm of pharmacological reprogramming of cells. Continuously advancing technologies in pharmacological reprogramming of cells may benefit and advance regenerative medicine. The established pharmacological tools have already been applied to enhance the processes of cellular reprogramming and improve the quality of cells for their clinical applications. The rapidly increasing number of readily available bioactive chemical tools will fuel our efforts to reprogram cells for transplantation therapies.
Collapse
Affiliation(s)
- Min Xie
- Gladstone Institutes, 1650 Owens Street, San Francisco, California 94158, United States
| | - Shibing Tang
- Gladstone Institutes, 1650 Owens Street, San Francisco, California 94158, United States
| | - Ke Li
- Gladstone Institutes, 1650 Owens Street, San Francisco, California 94158, United States
| | - Sheng Ding
- Gladstone Institutes, 1650 Owens Street, San Francisco, California 94158, United States
| |
Collapse
|
47
|
Clayton ZE, Yuen GS, Sadeghipour S, Hywood JD, Wong JW, Huang NF, Ng MK, Cooke JP, Patel S. A comparison of the pro-angiogenic potential of human induced pluripotent stem cell derived endothelial cells and induced endothelial cells in a murine model of peripheral arterial disease. Int J Cardiol 2017; 234:81-89. [DOI: 10.1016/j.ijcard.2017.01.125] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 12/28/2016] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
|
48
|
Zhang L, Jambusaria A, Hong Z, Marsboom G, Toth PT, Herbert BS, Malik AB, Rehman J. SOX17 Regulates Conversion of Human Fibroblasts Into Endothelial Cells and Erythroblasts by Dedifferentiation Into CD34 + Progenitor Cells. Circulation 2017; 135:2505-2523. [PMID: 28381471 PMCID: PMC5472005 DOI: 10.1161/circulationaha.116.025722] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 03/24/2017] [Indexed: 01/01/2023]
Abstract
Supplemental Digital Content is available in the text. Background: The mechanisms underlying the dedifferentiation and lineage conversion of adult human fibroblasts into functional endothelial cells have not yet been fully defined. Furthermore, it is not known whether fibroblast dedifferentiation recapitulates the generation of multipotent progenitors during embryonic development, which give rise to endothelial and hematopoietic cell lineages. Here we established the role of the developmental transcription factor SOX17 in regulating the bilineage conversion of fibroblasts by the generation of intermediate progenitors. Methods: CD34+ progenitors were generated after the dedifferentiation of human adult dermal fibroblasts by overexpression of pluripotency transcription factors. Sorted CD34+ cells were transdifferentiated into induced endothelial cells and induced erythroblasts using lineage-specific growth factors. The therapeutic potential of the generated cells was assessed in an experimental model of myocardial infarction. Results: Induced endothelial cells expressed specific endothelial cell surface markers and also exhibited the capacity for cell proliferation and neovascularization. Induced erythroblasts expressed erythroid surface markers and formed erythroid colonies. Endothelial lineage conversion was dependent on the upregulation of the developmental transcription factor SOX17, whereas suppression of SOX17 instead directed the cells toward an erythroid fate. Implantation of these human bipotential CD34+ progenitors into nonobese diabetic/severe combined immunodeficiency (NOD-SCID) mice resulted in the formation of microvessels derived from human fibroblasts perfused with mouse and human erythrocytes. Endothelial cells generated from human fibroblasts also showed upregulation of telomerase. Cell implantation markedly improved vascularity and cardiac function after myocardial infarction without any evidence of teratoma formation. Conclusions: Dedifferentiation of fibroblasts to intermediate CD34+ progenitors gives rise to endothelial cells and erythroblasts in a SOX17-dependent manner. These findings identify the intermediate CD34+ progenitor state as a critical bifurcation point, which can be tuned to generate functional blood vessels or erythrocytes and salvage ischemic tissue.
Collapse
Affiliation(s)
- Lianghui Zhang
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.)
| | - Ankit Jambusaria
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.)
| | - Zhigang Hong
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.)
| | - Glenn Marsboom
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.)
| | - Peter T Toth
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.)
| | - Brittney-Shea Herbert
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.)
| | - Asrar B Malik
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.)
| | - Jalees Rehman
- From Department of Pharmacology (L.Z., A.J., Z.H., G.M., P.T.T., A.B.M., J.R.), Department of Medicine, Division of Cardiology (J.R.), The University of Illinois College of Medicine, Chicago; and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis (B.-S.H.).
| |
Collapse
|
49
|
Cochrane A, Kelaini S, Tsifaki M, Bojdo J, Vilà-González M, Drehmer D, Caines R, Magee C, Eleftheriadou M, Hu Y, Grieve D, Stitt AW, Zeng L, Xu Q, Margariti A. Quaking Is a Key Regulator of Endothelial Cell Differentiation, Neovascularization, and Angiogenesis. Stem Cells 2017; 35:952-966. [PMID: 28207177 PMCID: PMC5396345 DOI: 10.1002/stem.2594] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/10/2017] [Accepted: 01/24/2017] [Indexed: 12/28/2022]
Abstract
The capability to derive endothelial cell (ECs) from induced pluripotent stem cells (iPSCs) holds huge therapeutic potential for cardiovascular disease. This study elucidates the precise role of the RNA‐binding protein Quaking isoform 5 (QKI‐5) during EC differentiation from both mouse and human iPSCs (hiPSCs) and dissects how RNA‐binding proteins can improve differentiation efficiency toward cell therapy for important vascular diseases. iPSCs represent an attractive cellular approach for regenerative medicine today as they can be used to generate patient‐specific therapeutic cells toward autologous cell therapy. In this study, using the model of iPSCs differentiation toward ECs, the QKI‐5 was found to be an important regulator of STAT3 stabilization and vascular endothelial growth factor receptor 2 (VEGFR2) activation during the EC differentiation process. QKI‐5 was induced during EC differentiation, resulting in stabilization of STAT3 expression and modulation of VEGFR2 transcriptional activation as well as VEGF secretion through direct binding to the 3′ UTR of STAT3. Importantly, mouse iPS‐ECs overexpressing QKI‐5 significantly improved angiogenesis and neovascularization and blood flow recovery in experimental hind limb ischemia. Notably, hiPSCs overexpressing QKI‐5, induced angiogenesis on Matrigel plug assays in vivo only 7 days after subcutaneous injection in SCID mice. These results highlight a clear functional benefit of QKI‐5 in neovascularization, blood flow recovery, and angiogenesis. Thus, they provide support to the growing consensus that elucidation of the molecular mechanisms underlying EC differentiation will ultimately advance stem cell regenerative therapy and eventually make the treatment of cardiovascular disease a reality. The RNA binding protein QKI‐5 is induced during EC differentiation from iPSCs. RNA binding protein QKI‐5 was induced during EC differentiation in parallel with the EC marker CD144. Immunofluorescence staining showing that QKI‐5 is localized in the nucleus and stained in parallel with CD144 in differentiated ECs (scale bar = 50 µm). stemcells2017 Stem Cells2017;35:952–966
Collapse
Affiliation(s)
- Amy Cochrane
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Sophia Kelaini
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Marianna Tsifaki
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - James Bojdo
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Marta Vilà-González
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Daiana Drehmer
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Rachel Caines
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Corey Magee
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Magdalini Eleftheriadou
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Yanhua Hu
- Cardiovascular Division, King's College London, London, United Kingdom
| | - David Grieve
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Alan W Stitt
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| | - Lingfang Zeng
- Cardiovascular Division, King's College London, London, United Kingdom
| | - Qingbo Xu
- Cardiovascular Division, King's College London, London, United Kingdom
| | - Andriana Margariti
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, United Kingdom
| |
Collapse
|
50
|
Sox17 drives functional engraftment of endothelium converted from non-vascular cells. Nat Commun 2017; 8:13963. [PMID: 28091527 PMCID: PMC5260855 DOI: 10.1038/ncomms13963] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/16/2016] [Indexed: 01/01/2023] Open
Abstract
Transplanting vascular endothelial cells (ECs) to support metabolism and express regenerative paracrine factors is a strategy to treat vasculopathies and to promote tissue regeneration. However, transplantation strategies have been challenging to develop, because ECs are difficult to culture and little is known about how to direct them to stably integrate into vasculature. Here we show that only amniotic cells could convert to cells that maintain EC gene expression. Even so, these converted cells perform sub-optimally in transplantation studies. Constitutive Akt signalling increases expression of EC morphogenesis genes, including Sox17, shifts the genomic targeting of Fli1 to favour nearby Sox consensus sites and enhances the vascular function of converted cells. Enforced expression of Sox17 increases expression of morphogenesis genes and promotes integration of transplanted converted cells into injured vessels. Thus, Ets transcription factors specify non-vascular, amniotic cells to EC-like cells, whereas Sox17 expression is required to confer EC function.
Endothelial cells (ECs) are promising strategies to treat vasculopathies but little is known about the factors that sustain EC identity and govern functional integration into vasculature after transplantation. Here the authors show that Ets factors and Sox17 convert nonvascular cells to vascular cells with stable EC identity and function.
Collapse
|